Abstract

Autophagy has been shown to play an essential role in defending against intracellular bacteria, viruses, and parasites. Mounting evidence suggests that autophagy plays different roles in the infection process of different pathogens. Until now, there has been no conclusive evidence regarding whether host autophagy is involved in Neospora caninum infection. In the current study, we first monitored the activation of autophagy by N. caninum, which occurred mainly in the early stages of infection, and examined the role of host autophagy in N. caninum infection. Here, we presented evidence that N. caninum induced an increase in autophagic vesicles with double-membrane structures in macrophages at the early stage of infection. LC3-II expression peaked and decreased as infection continued. However, the expression of P62/SQSTM1 showed significant accumulation within 12 h of infection, indicating that autophagic flux was blocked. A tandem fluorescence protein mCherry-GFP-LC3 construct was used to corroborate the impaired autophagic flux. Subsequently, we found that N. caninum infection induced the activation of the TLR2–AKT–mTOR pathways. Further investigation revealed that TLR2–mTOR, accompanied by the blockade of autophagic flux, was responsible for impaired autophagy but was not associated with AKT. In vitro and in vivo, N. caninum replication was strongly blocked by the kinase inhibitor 3-methyladenine (3-MA, autophagy inhibitor). In contrast, rapamycin (Rapa, an autophagy inducer) was able to promote intracellular proliferation and reduce the survival rate of N. caninum-infected mice. On the other hand, the accumulation of autophagosomes facilitated the proliferation of N. caninum. Collectively, our findings suggest that activation of host autophagy facilitates N. caninum replication and may counteract the innate immune response of the host. In short, inhibition of the early stages of autophagy could potentially be a strategy for neosporosis control.

Highlights

  • Neospora caninum (N. caninum), an intracellular protozoan parasite, is closely related to Toxoplasma gondii and causes abortion and reduced milk production in cattle, leading to financial losses worldwide (Reichel et al, 2013; Horcajo et al, 2016)

  • We found that N. caninum infection impairs TLR2–mTOR-dependent autophagy

  • To investigate whether autophagy could be involved during infection, peritoneal macrophages were infected with N. caninum and the autophagy level was determined

Read more

Summary

Introduction

Neospora caninum (N. caninum), an intracellular protozoan parasite, is closely related to Toxoplasma gondii and causes abortion and reduced milk production in cattle, leading to financial losses worldwide (Reichel et al, 2013; Horcajo et al, 2016). Evidence indicates that N. caninum infections have been detected in humans (Lobato et al, 2006; Duarte et al, 2020) Innate immune cells, such as macrophages, play a crucial role in controlling the initial parasite replication and pathogenesis of neosporosis, as these cells contribute to the first line of defense against intracellular infection. NF-kB, MAPK, and JAK/STAT signal pathways have been shown associated with infection (Jin et al, 2017; Nishikawa et al, 2018; Sharma et al, 2018) They influence the adaptive immune response by secreting many effector molecules including cytokines (Boucher et al, 2018; Jimenez-Pelayo et al, 2019; Miranda et al, 2019), controlling the proliferation and infection of N. caninum

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call